US20100311829A1 - Novel Therapeutic Uses of Beta-3 Adrenergic Receptor Agonist Derivatives in Particular to Modulate Apoptosis - Google Patents

Novel Therapeutic Uses of Beta-3 Adrenergic Receptor Agonist Derivatives in Particular to Modulate Apoptosis Download PDF

Info

Publication number
US20100311829A1
US20100311829A1 US12/652,317 US65231710A US2010311829A1 US 20100311829 A1 US20100311829 A1 US 20100311829A1 US 65231710 A US65231710 A US 65231710A US 2010311829 A1 US2010311829 A1 US 2010311829A1
Authority
US
United States
Prior art keywords
alkyl group
group
phenyl
alkyl
nhso
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US12/652,317
Other versions
US9161925B2 (en
Inventor
Marc BARDOU
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sanofi SA
Original Assignee
Sanofi Aventis France
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sanofi Aventis France filed Critical Sanofi Aventis France
Assigned to SANOFI-AVENTIS reassignment SANOFI-AVENTIS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BARDOU, MARC
Publication of US20100311829A1 publication Critical patent/US20100311829A1/en
Assigned to SANOFI reassignment SANOFI CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: SANOFI-AVENTIS
Application granted granted Critical
Publication of US9161925B2 publication Critical patent/US9161925B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/06Antiabortive agents; Labour repressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Definitions

  • the present invention relates to the novel therapeutic uses of beta-3 adrenergic receptor agonist derivatives in particular to modulate apoptosis.
  • Beta-2-adrenergic receptor (ADRB2) agonists have long been used as tocolytic agents for the treatment of preterm labor, but, due to limited efficacy and toxicity, are no longer recommended as first choice drugs.
  • Oxytocin antagonists and calcium channel blockers are more recently developed drugs with a lower profile of adverse affects, and hence more commonly used, even if calcium channel blockers, which were developed to treat hypertension, including in pregnant women, are not approved as tocolytic agents (Kim et al., Bjog 113 Suppl 3:113-115, 2006).
  • ADRB3 is present, and is the predominant ADRB, in human myometrium (Bardou et al., Br J Pharmacol 130:1960-1966, 2000; Dennedy et al., Bjog 108:605-609, 2001; Rouget et al., J Clin Endocrinol Metab 90:1644-1650, 2005) with an inhibitory action on spontaneous contractions.
  • Apoptosis is one of the main types of programmed cell death (PCD), and involves an orchestrated series of biochemical events leading to a characteristic cell morphology and death.
  • PCD programmed cell death
  • Research on apoptosis has increased substantially since the early 1990s.
  • defective apoptotic processes have been implicated in an extensive variety of diseases.
  • Cancer is a prime example of the crucial role of apoptotic regulation as defective anti-apoptotic effectors may result in uncontrolled cell proliferation.
  • Apoptosis is also largely involved in uterine cell turnover and early pregnancy. It is well-documented that the rodent uterine epithelium around the embryo undergoes apoptosis in response to the presence of the blastocyst (Schlafke et al., Anat. Rec. 212:47-56, 1985; Parr et al., Biol. Reprod. 36:211-225, 1987; and Welsh et al., Am. J. Anat. 192:215-231, 1991). Joswig et al., Reproductive Biology and Endocrinology 1:44, 2003, proposed two different pathways for apoptosis in uterine epithelium and decidua in response to implantation.
  • the first topic of interest for apoptosis in the field of pregnancy is the pathologies of the placenta. Indeed apoptosis of villous trophoblast is upregulated in both of the common pregnancy diseases related to the placenta, namely, intrauterine growth restriction (IUGR) and preeclampsia (Smith S C, et al. Am J Obstet Gynecol 1997; 177:1395-401; Allaire A D et al. Obstet Gynecol 2000; 96:271-6; Erel C T et al. Int J Gynaecol Obstet 2001; 73:229-35; Ishihara N, et al.
  • IUGR intrauterine growth restriction
  • Spontaneous preterm labor is one of the largest causes of preterm birth which in turn is the most frequent cause of infant death in the United States (Callaghan et al., Pediatrics 118, 1566-1573, 2006). Many cases of spontaneous preterm labor are unexplained but a significant proportion is linked to genital tract infection or chorioamnionitis (Edwards et al., Obstet. Gynecol. Clin. North Am. 32, 287-296, 2005).
  • Apoptotic cell death can be initiated by two alternative convergent pathways: the extrinsic pathway, which is mediated by cell surface death receptors, and the intrinsic pathway, which is mediated by mitochondria (Danial et al., Cell 116:205-219, 2004).
  • the extrinsic pathway which is mediated by cell surface death receptors
  • the intrinsic pathway which is mediated by mitochondria
  • cysteine aspartyl-specific proteases which cleave cellular substrates are activated
  • activation of the effector caspase-3 is important for the execution of apoptotic cell death.
  • the bcl2 family members play a central role in the regulation of apoptosis.
  • WO02/44139 discloses that certain propanolamines bearing a cyclohexyl(alkyl) group on the amine possess a powerful agonist activity with respect to ADRB3.
  • these compounds may be indicated in the treatment of gastrointestinal diseases such as inflammatory diseases of the intestine, for instance irritable bowel disease (IBD), as modulators of intestinal motivity, as lipolytic agents, anti-obesity agents, anti-diabetic agents, psychotropic agents, anti-glaucoma agents, cicatrizing agents and anti-depressants, as inhibitors of uterine contractions, as tocolytics for preventing or delaying preterm births, and for the treatment and/or prophylaxis of dysmenorrhoea.
  • IBD irritable bowel disease
  • these compounds could be used in the treatment of certain diseases of the central nervous system, such as for example depression, and also of certain disorders of the urinary system, such as urinary incontinence.
  • ADRB3 plays a crucial role in controlling apoptotic pathways, in particular in pregnancy related tissues, namely uterus, placenta and fetal membranes. Additionally, it has also been surprisingly discovered that compounds disclosed in WO02/44139 are potent apoptosis inhibitors.
  • the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament for preventing pregnancy related tissues apoptosis in female mammalian patients.
  • the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or treat pregnancy related tissues apoptosis related diseases and/or disorders; more particularly to prevent and/or treat uterine apoptosis related diseases and/or disorders; more particularly to prevent and/or treat pregnancy related tissues apoptosis related diseases and/or disorders selected from sexual cycle disorders, fertility disorders and pregnancy disorders.
  • the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or treat pregnancy related tissues apoptosis related diseases and/or disorders in female mammalian patients suffering from genital tract infection; more particularly to prevent and/or treat preterm labor in female mammalian patients suffering from genital tract infection; more particularly to prevent and/or treat preterm labor in female mammalian patients suffering from genital tract infection wherein genital tract infection is chorioamnionitis.
  • the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or inhibit pregnancy related tissues apoptosis in pregnant women.
  • the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament for preventing uterine apoptosis in women suffering from genital tract infection, with its consequences mainly preterm labor and preterm delivery.
  • the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or treat fetal membranes apoptosis in women suffering from preterm premature rupture of fetal membranes.
  • the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or treat placental apoptosis in women with pregnancy complicated by intrauterine growth restriction or preeclampsia.
  • beta-3 adrenergic receptor agonist is selected from compounds of formula (I):
  • A is a group of formula (a) or (b):
  • R represents a hydrogen or halogen atom, an —S(O) z (C 1 -C 4 )alkyl group, an —NHSO 2 (C 1 -C 4 )alkyl group, an —SO 2 NH(C 1 -C 4 )alkyl group, an —NHSO 2 -phenyl-(C 1 -C 4 )alkyl group or an —NHSO 2 -phenyl group, said phenyl possibly being substituted with a halogen atom, with a (C 1 -C 4 )alkyl group or with a (C 1 -C 4 )alkoxy group;
  • R 1 represents a hydrogen atom or a (C 1 -C 4 )alkyl group, a —CO(C 1 -C 4 )alkyl group, a phenyl-(C 1 -C 4 )alkyl group or a —CO-phenyl group, said phenyl possibly being substituted with a halogen atom or with a (C 1 -C 4 )alkoxy group;
  • R 2 is a hydrogen atom, an —SO 2 (C 1 -C 4 )alkyl group, an —SO 2 -phenyl-(C 1 -C 4 )alkyl group or an —SO 2 -phenyl group;
  • X completes a ring of 5 to 8 atoms, said ring being saturated or unsaturated, possibly being substituted with one or two (C 1 -C 4 )alkyl groups and bearing one or two carbonyl groups;
  • n, m and z are, independently, 0, 1 or 2;
  • R3 represents a hydrogen or halogen atom, a (C 1 -C 6 )alkyl group, a (C 1 -C 4 )alkoxy group, a —COO(C 1 -C 4 )alkyl group, a —CO(C 1 -C 4 )alkyl group, an —NHSO 2 (C 1 -C 4 )alkyl group, an —NHSO 2 -phenyl-(C 1 -C 4 )alkyl group, —NO 2 , —CN, —CONR 4 R 5 , —COOH, or a 4,5-dihydro-1,3-oxazol-2-yl or 4,4-dimethyl-4,5-dihydro-1,3-oxazol-2-yl group;
  • R 4 and R 5 represent, independently, a hydrogen atom, a phenyl, a (C 1 -C 4 )alkyl group or a phenyl-(C 1 -C 4 )alkyl group; or
  • R 4 and R 5 with the nitrogen atom to which they are attached, may form a ring of 5 to 7 atoms in total;
  • (C 1 -C 4 )alkyl and “(C 1 -C 6 )alkyl” denote monovalent radicals formed from a respectively C 1 -C 4 and C 1 -C 6 hydrocarbon containing a straight or branched saturated chain.
  • halogen denotes an atom chosen from chlorine, bromine, iodine and fluorine.
  • More particularly compounds are those in which n and m are each zero.
  • More particularly compounds are those in which R 1 is a hydrogen atom.
  • More particularly compounds are those in which R is chosen from an —NHSO 2 (C 1 -C 4 )alkyl group, an —NHSO 2 -phenyl-(C 1 -C 4 )alkyl group or an —NHSO 2 -phenyl group.
  • R 3 is —COO(C 1 -C 4 )alkyl or —CO(C 1 -C 4 )alkyl or CONR 4 R 5 .
  • More particularly compounds are those in which R 3 is in position 4 of the benzene.
  • More particularly compounds are those in which z is 2.
  • More particularly compounds are those in which X is a methylene, an ethylene or a propylene.
  • More particularly compounds are those in which X is a carbonyl, a —CO—CO-group, a —CO—C((C 1 -C 4 )alkyl) 2 -CO— group, a methylene monosubstituted or disubstituted with (C 1 -C 4 )alkyl or a —COCH 2 — group.
  • —NHSO 2 -phenyl-(C 1 -C 4 )alkyl and —SO 2 -phenyl-(C 1 -C 4 )alkyl groups are, respectively, benzylsulphonylamino and benzylsulphonyl.
  • R 4 and R 5 form, with the nitrogen atom to which they are attached, a ring of 5 to 7 atoms, rings more particularly are piperidine and pyrrolidine.
  • the salts of the compounds of formula (I) according to the present invention comprise both the addition salts with pharmaceutically acceptable inorganic or organic acids, such as hydrochlorate, hydrobromate, sulphate, hydrogen sulphate, dihydrogen phosphate, citrate, maleate, tartrate, fumarate, gluconate, methanesulphonate, 2-naphtalenesulphonate, etc., and the addition salts which allow suitable separation or crystallization of the compounds of formula (I), such as picrate or oxalate, or the addition salts with optically active acids, for example camphorsulphonic acids and mandelic or substituted mandelic acids.
  • pharmaceutically acceptable inorganic or organic acids such as hydrochlorate, hydrobromate, sulphate, hydrogen sulphate, dihydrogen phosphate, citrate, maleate, tartrate, fumarate, gluconate, methanesulphonate, 2-naphtalenesulphonate, etc.
  • the salts also comprise the salts with inorganic bases, preferably those with alkaline metals such as sodium or potassium, or with organic bases.
  • optically pure stereoisomers and also the mixtures of isomers of the compounds of formula (I), due to the asymmetric carbon, in any proportion, are also part of the present invention.
  • More particularly compounds of formula (I) are the compounds in which the configuration of the carbon of the propanolamine bearing the OH group is (S).
  • the compounds of formula (I) may be in the form of “cis” or “trans” geometrical isomers, depending on the relative position of the substituents in positions 1 and 4 of the cyclohexyl ring (marked with a star). These pure isomers and their mixtures, in any proportion, are part of the present invention.
  • the compounds of formula (I) are useful for the preparation of medicaments suitable to prevent and/or treat, in female patients in need thereof, diseases and/or disorders related to pregnancy related tissues apoptosis related, such as sexual cycle disorders, fertility disorders and pregnancy disorders, as well as preterm premature rupture of fetal membranes, as well as preterm labor, in particular preterm labor triggered by genital tract infection.
  • diseases and/or disorders related to pregnancy related tissues apoptosis related such as sexual cycle disorders, fertility disorders and pregnancy disorders, as well as preterm premature rupture of fetal membranes, as well as preterm labor, in particular preterm labor triggered by genital tract infection.
  • diseases and/or disorders related to pregnancy related tissues apoptosis related such as sexual cycle disorders, fertility disorders and pregnancy disorders, as well as preterm premature rupture of fetal membranes, as well as preterm labor, in particular preterm labor triggered by genital tract infection.
  • preterm labor in particular preterm labor triggered by genital tract infection.
  • female mammalian patients refers in particular to women.
  • fertility disorders refers to disorders of the female fertility, in particular those associated with impaired uterine apoptosis, such as implantation as disclosed by Schlafke et al., Anat. Rec. 212:47-56, 1985; Parr et al., Biol. Reprod. 36:211-225, 1987; Welsh et al., Am. J. Anat. 192:215-231, 1991 and Joswig et al., Reproductive Biology and Endocrinology 1:44, 2003.
  • pregnancy disorders refers to disorders of the pregnancy in female mammalian, in particular those associated with impaired uterine apoptosis, such as early pregnancy as as disclosed by Schlafke et al., Anat. Rec. 212:47-56, 1985; Parr et al., Biol. Reprod. 36:211-225, 1987; Welsh et al., Am. J. Anat. 192:215-231, 1991 and Joswig et al., Reproductive Biology and Endocrinology 1:44, 2003.
  • an effective amount of a compound of formula (I), or of a pharmaceutically acceptable salt thereof, is administered to the mammals which require such a treatment.
  • the compounds of formula (I) above, and the pharmaceutically acceptable salts thereof, may be used at daily doses of 0.01 to 20 mg per kilo of body weight of the mammal to be treated, preferably at daily doses of from 0.1 to 10 mg/kg.
  • the dose may vary preferably from 0.5 mg to 1500 mg per day, in particular from 2.5 to 500 mg, depending on the age of the individual to be treated, the type of treatment, prophylactic or curative, and the seriousness of the disorder.
  • the compounds of formula (I) are generally administered as a dosage unit of 0.1 to 500 mg, preferably of 0.5 to 100 mg, of active principle, one to five times a day.
  • Said dosage units are preferably formulated in pharmaceutical compositions in which the active principle is mixed with a pharmaceutical excipient.
  • the present invention relates to pharmaceutical compositions containing, as an active principle, a compound of formula (I) above or a pharmaceutically acceptable salt thereof.
  • the active ingredients of formula (I) above, and the pharmaceutically acceptable salts thereof may be administered in unit administration forms, mixed with conventional pharmaceutical supports, to animals and humans for treating the abovementioned disorders.
  • the unit administration forms which are suitable comprise oral forms such as tablets, gel capsules, powders, granules and oral solutions or suspensions, sublingual and buccal administration forms, subcutaneous, intramuscular or intravenous administration forms, local administration forms and rectal administration forms.
  • the main active ingredient is mixed with a pharmaceutical vehicle such as gelatin, starch, lactose, magnesium stearate, talc, gum arabic or the like.
  • a pharmaceutical vehicle such as gelatin, starch, lactose, magnesium stearate, talc, gum arabic or the like.
  • the tablets may be coated with sucrose or other suitable materials, or they may be treated such that they have sustained or delayed activity and that they release, in a continuous manner, a predetermined amount of active principle.
  • a preparation of gel capsules is obtained by mixing the active ingredient with a diluent and pouring the mixture obtained into soft or hard gel capsules.
  • a preparation in the form of a syrup or elixir may contain the active ingredient together with a sweetener, preferably a calorie-free sweetener, methylparaben and propylparaben as antiseptics, and also a flavour enhancer and a suitable colorant.
  • a sweetener preferably a calorie-free sweetener, methylparaben and propylparaben as antiseptics, and also a flavour enhancer and a suitable colorant.
  • the water-dispersible powders or granules may contain the active ingredient mixed with dispersing agents or wetting agents, or suspending agents, such as polyvinylpyrrolidone, and also with sweeteners or flavour correctors.
  • the active principle is mixed into an excipient for preparing creams or ointments, or it is dissolved in a vehicle for intraocular administration, for example in the form of an eyewash.
  • suppositories prepared with binders which melt at rectal temperature, for example cocoa butter or polyethylene glycols.
  • aqueous suspensions, saline solutions or injectable sterile solutions which contain pharmacologically compatible dispersion agents and/or wetting agents, for example propylene glycol or butylene glycol, are used.
  • the active principle may also be formulated in the form of microcapsules, optionally with one or more supports or additives.
  • FIG. 1 represents Western Blot Analysis with semi-quantification showing the expression of cleaved caspase-3 protein.
  • CA group myometrial samples obtained from 3 different women with Chorioamnionitis (CA group), and with 5 myometrium, obtained at cesarean delivery from 5 different women with uncomplicated pregnancies and stimulated, or not, with LPS (LPS and CTRL groups respectively). *P ⁇ 0.05 vs. controls.
  • FIG. 2 illustrates Western Blot Analysis with semi-quantification showing the expression of cleaved caspase-3 protein.
  • FIG. 3 illustrates Western Blot Analysis with semi-quantification showing the expression of cleaved caspase-3 protein.
  • FIG. 4 illustrates Western Blot Analysis with semi-quantification showing the expression of BAX and Bcl-2 protein.
  • FIG. 5 show the effect of Compound A on the concentrations of IL-6 and IL-8.
  • Results (mean ⁇ s.e.m, in pg/ml) are from five experiments performed in duplicate in myometrium, obtained at elective cesarean delivery, from five different women. #P ⁇ 0.05 vs controls, *P ⁇ 0.05 vs LPS, **P ⁇ 0.01 vs LPS.
  • Lipopolysaccharide ( Escherichia coli 055:B5, ref: L2880) was purchased from Sigma-Aldrich and was dissolved in distilled water.
  • Compound A was a gift from Sanofi-Midy Research Center, Exploratory Research Department, Sanofi-Aventis S.p.A. It was dissolved in a mixture of absolute ethanol 30%, dimethylsulfoxide 2%, and distilled water for the 1 mM solution and thereafter diluted in distilled water. The final maximal bath concentration was 0.3% for ethanol and 0.02% for DMSO.
  • Myometrial biopsies were obtained from women during pregnancy in four different clinical situations as outlined: i) a woman with established chorioamnionitis undergoing postpartum hysterectomy; ii) a woman undergoing postpartum hysterectomy for postpartum hemorrhage in the absence of chorioamnionitis; iii) 3 women undergoing elective caesarean section with established chorioamnionitis; iv) 10 women undergoing elective caesarean section for other reasons (cephalo-pelvic disproportion) in the absence of chorioamnionitis.
  • chorioamnionitis refers to confirmed chorioamnionitis.
  • Myometrial strips were excised from an immediately subserosal site where the majority of the fibers are in a longitudinal orientation, at an antiplacental site, as previously described (Rouget C, et al. J Clin Endocrinol Metab 2005; 90: 1644-1650, Leroy M J, et al. Biochem Pharmacol 1989; 38: 9-15). This study was approved by the “Comitati Consultatif de Protection des Personnes pour la mecanic Biometriccale” (CCPPRB, Dijon, France) and informed consent was obtained from all donors.
  • Tissues obtained from women with confirmed chorioamnionitis or postpartum hemorrhage were either used fresh to perform western blot experiments, or embedded in paraffin for histological assessment and Hoechst staining, as described below.
  • Myometrial biopsies obtained from uncomplicated pregnancies were used to develop the LPS experimental model mimicking the effects of chorioamnionitis.
  • the supernatants samples and tissues were quickly frozen in liquid nitrogen and stored at ⁇ 80° C.
  • Proteins were transferred to a nitrocellulose membrane (Hybond-P, Amersham Biosciences).
  • a nitrocellulose membrane Hybond-P, Amersham Biosciences.
  • membranes were incubated for 1 h in 10% nonfat dried milk powder in Tris-buffered saline/Tween 20 (TBST) (10 mM Tris, 150 mM NaCl, and 0.1% Tween 20, pH 7.8) at room temperature. Blocked membranes were washed 3 times with TBST. The blots were then incubated overnight at 4° C.
  • TST Tris-buffered saline/Tween 20
  • the intensities of the bands were analyzed densitometrically using the NIH Image 1.62 program and normalized with intensity of bands obtained with monoclonal antibody to Glyceraldehyde 3-Phosphate Dehydrogenase (GAPDH) used as protein loading control. Results are expressed as the mean ⁇ s.e.m. in Arbitrary Density Unit (A.D.U).
  • the myometrial strips were fixed for 1 h with paraformaldehyde 4%, then embedded in paraffin and cut into 5-micrometer-thick sections.
  • antigen retrieval was performed by incubating slides 10 min in warm citric acid buffer pH 6 with a pressure cooker. After endogenous peroxidase activity was blocked with hydrogen peroxide (H 2 O 2 ) 3%, slides were incubated either with primary anti-cleaved caspase-3 IgG (1:100), washed three times with phosphate buffered saline (PBS) and then incubated with biotinylated anti-rabbit (1:600) immunoglobulin respectively for 1 h.
  • PBS phosphate buffered saline
  • chromatin condensation state was assessed by staining of nuclei with Hoechst-33342. Paraffin embedded sections of myometrium samples were re-hydrated as described above. Slides were then incubated for 2 min with Hoechst-33342 (2 ⁇ g/ml). The slides were rinsed with distilled water and mounted with Aquatex ⁇ . The stained nuclei were visualized with convert fluorescent microscope at a magnification of ⁇ 100, using excitation light at 350 nm.
  • PCR analyses were performed using 25 ng of reverse transcribed total RNA with 200 nM of both sense and anti sense primers in a final volume of 25 ⁇ l using the SYBR Green PCR jumpstart reagent (Sigma, Saint Louis, Mo., USA) in an iCycler iQ real time detection system instrument (Bio-Rad, Marnes-1a-Coquette, France). PCR products were also analyzed on edithium bromide stained agarose gel to ensure that a single amplicon of the expected size was indeed obtained.
  • Glyceraldehyde-3-phosphate dehydrogenase Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used in each experiment to control for variability in the quantities of cDNA.
  • ⁇ Ct Ct of the experimental group—Ct of the control group.
  • PCR was performed using specific primers: GAPDH (Forward, 5′TGCACCACCAACTGCTTAGC3′ (SEQ ID NO:1), and Reverse, 5′GGCATGGACTGTGGTC-ATGAG3′ (SEQ ID NO:2)) and CASPASE-3 (Forward, 5′ AGAACTGGACTGTGGCATTGAG 3′ (SEQ ID NO:3), and Reverse, 5′ GCTTGTCGGCATACTGTTTCAG 3′ (SEQ ID NO:4)).
  • Cytokines measurements were determined by Cytometric Bead Array (CBA; Bender MedSystems). Supernatants of 5 myometrial strips were incubated with labeled capture beads and detection reagent for 3 h in the dark at room temperature, and analyzed with a flow cytometer (FACSCalibur; BD Biosciences) by using the respective CBA Analysis software (BD Biosciences) and Bender MedSystems software. Cytokine standards for quantification (pg/mL) as well as the blanks were handled in the same manner as the samples. Each experiment was performed in duplicate to ensure for reproducibility of results.
  • CBA Cytometric Bead Array
  • LPS stimulation in optimal conditions i.e 10 ⁇ g/ml for 48 h, determined after time course—8 h, 24 h, 48 h—and dose response—50 ng/ml, 1 ⁇ g/ml, 10 ⁇ g/ml—experiments of LPS stimulation
  • dose response 50 ng/ml, 1 ⁇ g/ml, 10 ⁇ g/ml—experiments of LPS stimulation
  • the solvent for the highest concentration of Compound A i.e. distilled water containing ethanol 0.3% and DMSO 0.02%, had no effect on cleaved caspase-3, BAX and BCL2 expression in tissues either not stimulated or stimulated with LPS (data not shown).
  • a unitary dosage form of a compound of the invention in the form of a tablet may comprise the following constituents:

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Reproductive Health (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Cardiology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to the new therapeutic uses of beta-3 adrenergic receptor agonist derivatives in particular to inhibit apoptosis and treat and/or prevent apoptosis-related diseases and disorders.

Description

  • The present invention relates to the novel therapeutic uses of beta-3 adrenergic receptor agonist derivatives in particular to modulate apoptosis.
  • Beta-2-adrenergic receptor (ADRB2) agonists have long been used as tocolytic agents for the treatment of preterm labor, but, due to limited efficacy and toxicity, are no longer recommended as first choice drugs. Oxytocin antagonists and calcium channel blockers are more recently developed drugs with a lower profile of adverse affects, and hence more commonly used, even if calcium channel blockers, which were developed to treat hypertension, including in pregnant women, are not approved as tocolytic agents (Kim et al., Bjog 113 Suppl 3:113-115, 2006). It has recently been shown that ADRB3 is present, and is the predominant ADRB, in human myometrium (Bardou et al., Br J Pharmacol 130:1960-1966, 2000; Dennedy et al., Bjog 108:605-609, 2001; Rouget et al., J Clin Endocrinol Metab 90:1644-1650, 2005) with an inhibitory action on spontaneous contractions.
  • Apoptosis is one of the main types of programmed cell death (PCD), and involves an orchestrated series of biochemical events leading to a characteristic cell morphology and death. Research on apoptosis has increased substantially since the early 1990s. In addition to its importance as a biological phenomenon, defective apoptotic processes have been implicated in an extensive variety of diseases. Cancer is a prime example of the crucial role of apoptotic regulation as defective anti-apoptotic effectors may result in uncontrolled cell proliferation.
  • The review by Solary et al., Eur. Respir. J. 9, 1293-1305, 1996 confirms the role of apoptosis in cancers; viral infections; autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis, autoimmune diabetes mellitus, inflammatory bowel syndrome, type 1 diabetes mellitus; fas-mediated disease such as fulminent hepatitis, CTL-mediated autoimmune disease including chronic thyroiditis; AIDS; haematological diseases such as myelodisplastic syndromes, aplastic anaemia, chronic neutropenia, severe beta-thalassaemia; neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, cerebellar degeneration, retinal pigmentosa, spinal muscular atrophy; and other various diseases including polycystic kidney disease, toxic-induced liver diseases, ischaemic injury, such as myocardial infarction or stroke, atheroma, arthritis, osteoporosis and ageing.
  • Apoptosis is also largely involved in uterine cell turnover and early pregnancy. It is well-documented that the rodent uterine epithelium around the embryo undergoes apoptosis in response to the presence of the blastocyst (Schlafke et al., Anat. Rec. 212:47-56, 1985; Parr et al., Biol. Reprod. 36:211-225, 1987; and Welsh et al., Am. J. Anat. 192:215-231, 1991). Joswig et al., Reproductive Biology and Endocrinology 1:44, 2003, proposed two different pathways for apoptosis in uterine epithelium and decidua in response to implantation. More recently, Qian Zhang et al., Endocrinology 147(5), 2215-2227, 2006, studied apoptosis and proliferation in uterine cell turnover during the estrous cycle and early pregnancy. Their results indicate that uterine cell apoptosis and proliferation patterns are highly ordered cell-specific phenomena that play an important role in maintaining the sexual cycle and pregnancy-associated uterine changes. In particular, they indicate that caspase-3-mediated cell apoptosis may play an important role in initiating the implantation process in hamsters and mice.
  • The first topic of interest for apoptosis in the field of pregnancy is the pathologies of the placenta. Indeed apoptosis of villous trophoblast is upregulated in both of the common pregnancy diseases related to the placenta, namely, intrauterine growth restriction (IUGR) and preeclampsia (Smith S C, et al. Am J Obstet Gynecol 1997; 177:1395-401; Allaire A D et al. Obstet Gynecol 2000; 96:271-6; Erel C T et al. Int J Gynaecol Obstet 2001; 73:229-35; Ishihara N, et al. Am J Obstet Gynecol 2002; 186:158-66.). Since it has recently been shown that apoptotic nuclei are more abundant in fetal growth restricted placentas compared with control placentas (Madazli R, et al. J Obstet Gynaecol 2006; 26:5-10) inhibiting apoptosis can be considered as promising approach to treat or prevent IUGR or preeclampsia.
  • Spontaneous preterm labor, whether explained or unexplained, is one of the largest causes of preterm birth which in turn is the most frequent cause of infant death in the United States (Callaghan et al., Pediatrics 118, 1566-1573, 2006). Many cases of spontaneous preterm labor are unexplained but a significant proportion is linked to genital tract infection or chorioamnionitis (Edwards et al., Obstet. Gynecol. Clin. North Am. 32, 287-296, 2005).
  • Charpigny et al., Biol. Reprod., 68, 2289-2296, 2003, showed that apoptosis associated genes were up-regulated during parturition.
  • Apoptotic cell death can be initiated by two alternative convergent pathways: the extrinsic pathway, which is mediated by cell surface death receptors, and the intrinsic pathway, which is mediated by mitochondria (Danial et al., Cell 116:205-219, 2004). In both pathways, cysteine aspartyl-specific proteases (caspases) which cleave cellular substrates are activated, and activation of the effector caspase-3 is important for the execution of apoptotic cell death. The bcl2 family members play a central role in the regulation of apoptosis. The multidomain proapoptotic proteins Bax and Bak together constitute a requisite gateway to apoptotic cell death because cells doubly deficient for these proteins are resistant to several different intrinsic death stimuli (Danial et al., Cell 116:205-219, 2004). Therefore BAX, Bcl-2 and cleaved caspase-3 are widely used to assess apoptosis.
  • WO02/44139 discloses that certain propanolamines bearing a cyclohexyl(alkyl) group on the amine possess a powerful agonist activity with respect to ADRB3. This document suggests that these compounds may be indicated in the treatment of gastrointestinal diseases such as inflammatory diseases of the intestine, for instance irritable bowel disease (IBD), as modulators of intestinal motivity, as lipolytic agents, anti-obesity agents, anti-diabetic agents, psychotropic agents, anti-glaucoma agents, cicatrizing agents and anti-depressants, as inhibitors of uterine contractions, as tocolytics for preventing or delaying preterm births, and for the treatment and/or prophylaxis of dysmenorrhoea. In addition, these compounds could be used in the treatment of certain diseases of the central nervous system, such as for example depression, and also of certain disorders of the urinary system, such as urinary incontinence.
  • Nevertheless, the use of these compounds for preventing and/or inhibiting apoptosis is neither disclosed nor suggested. More generally the nexus between ADRB3 and apoptosis has not been established or suggested so far.
  • There is therefore a need to identify new biological targets for controlling apoptosis and modulators thereof.
  • The present inventors have now found, and that is an object of the present invention, that ADRB3 plays a crucial role in controlling apoptotic pathways, in particular in pregnancy related tissues, namely uterus, placenta and fetal membranes. Additionally, it has also been surprisingly discovered that compounds disclosed in WO02/44139 are potent apoptosis inhibitors.
  • According to a first object, the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament for preventing pregnancy related tissues apoptosis in female mammalian patients.
  • According to another object, the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or treat pregnancy related tissues apoptosis related diseases and/or disorders; more particularly to prevent and/or treat uterine apoptosis related diseases and/or disorders; more particularly to prevent and/or treat pregnancy related tissues apoptosis related diseases and/or disorders selected from sexual cycle disorders, fertility disorders and pregnancy disorders.
  • According to another object, the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or treat pregnancy related tissues apoptosis related diseases and/or disorders in female mammalian patients suffering from genital tract infection; more particularly to prevent and/or treat preterm labor in female mammalian patients suffering from genital tract infection; more particularly to prevent and/or treat preterm labor in female mammalian patients suffering from genital tract infection wherein genital tract infection is chorioamnionitis.
  • According to another object, the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or inhibit pregnancy related tissues apoptosis in pregnant women.
  • According to another object, the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament for preventing uterine apoptosis in women suffering from genital tract infection, with its consequences mainly preterm labor and preterm delivery.
  • According to another object, the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or treat fetal membranes apoptosis in women suffering from preterm premature rupture of fetal membranes.
  • According to another object, the present invention concerns the use of a beta-3 adrenergic receptor agonist for the preparation of a medicament suitable to prevent and/or treat placental apoptosis in women with pregnancy complicated by intrauterine growth restriction or preeclampsia.
  • According to another object, said beta-3 adrenergic receptor agonist is selected from compounds of formula (I):
  • Figure US20100311829A1-20101209-C00001
  • where in formula (I):
  • A is a group of formula (a) or (b):
  • Figure US20100311829A1-20101209-C00002
  • where:
  • R represents a hydrogen or halogen atom, an —S(O)z(C1-C4)alkyl group, an —NHSO2(C1-C4)alkyl group, an —SO2NH(C1-C4)alkyl group, an —NHSO2-phenyl-(C1-C4)alkyl group or an —NHSO2-phenyl group, said phenyl possibly being substituted with a halogen atom, with a (C1-C4)alkyl group or with a (C1-C4)alkoxy group;
  • R1 represents a hydrogen atom or a (C1-C4)alkyl group, a —CO(C1-C4)alkyl group, a phenyl-(C1-C4)alkyl group or a —CO-phenyl group, said phenyl possibly being substituted with a halogen atom or with a (C1-C4)alkoxy group;
  • R2 is a hydrogen atom, an —SO2(C1-C4)alkyl group, an —SO2-phenyl-(C1-C4)alkyl group or an —SO2-phenyl group;
  • X completes a ring of 5 to 8 atoms, said ring being saturated or unsaturated, possibly being substituted with one or two (C1-C4)alkyl groups and bearing one or two carbonyl groups;
  • n, m and z are, independently, 0, 1 or 2;
  • R3 represents a hydrogen or halogen atom, a (C1-C6)alkyl group, a (C1-C4)alkoxy group, a —COO(C1-C4)alkyl group, a —CO(C1-C4)alkyl group, an —NHSO2(C1-C4)alkyl group, an —NHSO2-phenyl-(C1-C4)alkyl group, —NO2, —CN, —CONR4R5, —COOH, or a 4,5-dihydro-1,3-oxazol-2-yl or 4,4-dimethyl-4,5-dihydro-1,3-oxazol-2-yl group;
  • R4 and R5 represent, independently, a hydrogen atom, a phenyl, a (C1-C4)alkyl group or a phenyl-(C1-C4)alkyl group; or
  • R4 and R5 with the nitrogen atom to which they are attached, may form a ring of 5 to 7 atoms in total;
  • and the salts thereof.
  • In the present description, the terms “(C1-C4)alkyl” and “(C1-C6)alkyl” denote monovalent radicals formed from a respectively C1-C4 and C1-C6 hydrocarbon containing a straight or branched saturated chain.
  • In the present description, the term “halogen” denotes an atom chosen from chlorine, bromine, iodine and fluorine.
  • More particularly compounds are those in which n and m are each zero.
  • More particularly compounds are those in which R1 is a hydrogen atom.
  • More particularly compounds are those in which R is chosen from an —NHSO2(C1-C4)alkyl group, an —NHSO2-phenyl-(C1-C4)alkyl group or an —NHSO2-phenyl group.
  • More particularly compounds are those in which R3 is —COO(C1-C4)alkyl or —CO(C1-C4)alkyl or CONR4R5.
  • More particularly compounds are those in which R3 is in position 4 of the benzene.
  • More particularly compounds are those in which z is 2.
  • More particularly compounds are those in which X is a methylene, an ethylene or a propylene.
  • More particularly compounds are those in which X is a carbonyl, a —CO—CO-group, a —CO—C((C1-C4)alkyl)2-CO— group, a methylene monosubstituted or disubstituted with (C1-C4)alkyl or a —COCH2— group.
  • More particularly —NHSO2-phenyl-(C1-C4)alkyl and —SO2-phenyl-(C1-C4)alkyl groups are, respectively, benzylsulphonylamino and benzylsulphonyl.
  • When R4 and R5 form, with the nitrogen atom to which they are attached, a ring of 5 to 7 atoms, rings more particularly are piperidine and pyrrolidine.
  • The following compound is particularly advantageous:
  • Ethyl-4-{trans-4-[((2S)-2-hydroxy-3-{4-hydroxy-3[(methylsulfonyl)-amino]phenoxy}propyl)amino]cyclohexyl}benzoate hydrochloride, herein called Compound A; it is a compound of formula (I) as disclosed above where A is a group of formula (a) where R is —NHSO2-Methyle, R1 is OH, n=m=0 and R3 is —COOEthyle.
  • The salts of the compounds of formula (I) according to the present invention comprise both the addition salts with pharmaceutically acceptable inorganic or organic acids, such as hydrochlorate, hydrobromate, sulphate, hydrogen sulphate, dihydrogen phosphate, citrate, maleate, tartrate, fumarate, gluconate, methanesulphonate, 2-naphtalenesulphonate, etc., and the addition salts which allow suitable separation or crystallization of the compounds of formula (I), such as picrate or oxalate, or the addition salts with optically active acids, for example camphorsulphonic acids and mandelic or substituted mandelic acids.
  • When the compounds of formula (I) have a free carboxyl group, the salts also comprise the salts with inorganic bases, preferably those with alkaline metals such as sodium or potassium, or with organic bases.
  • The optically pure stereoisomers, and also the mixtures of isomers of the compounds of formula (I), due to the asymmetric carbon, in any proportion, are also part of the present invention.
  • More particularly compounds of formula (I) are the compounds in which the configuration of the carbon of the propanolamine bearing the OH group is (S).
  • The compounds of formula (I) may be in the form of “cis” or “trans” geometrical isomers, depending on the relative position of the substituents in positions 1 and 4 of the cyclohexyl ring (marked with a star). These pure isomers and their mixtures, in any proportion, are part of the present invention.
  • The mixtures of optical and geometrical stereoisomers above, in any proportion, are also part of the present invention.
  • The process of preparation of the compounds of formula (I) is disclosed in WO 02/44139.
  • The compounds of formula (I) are useful for the preparation of medicaments suitable to prevent and/or treat, in female patients in need thereof, diseases and/or disorders related to pregnancy related tissues apoptosis related, such as sexual cycle disorders, fertility disorders and pregnancy disorders, as well as preterm premature rupture of fetal membranes, as well as preterm labor, in particular preterm labor triggered by genital tract infection. Such patients can be readily identified by the routine analysis.
  • The expression “female mammalian patients” used herein refers in particular to women.
  • The expression “sexual cycle disorders” used herein refers to disorders of the female menstrual cycle, in particular those associated with impaired uterine apoptosis, as suggested by Qian Zhang et al., Endocrinology 147(5), 2215-2227, 2006
  • The expression “fertility disorders” used herein refers to disorders of the female fertility, in particular those associated with impaired uterine apoptosis, such as implantation as disclosed by Schlafke et al., Anat. Rec. 212:47-56, 1985; Parr et al., Biol. Reprod. 36:211-225, 1987; Welsh et al., Am. J. Anat. 192:215-231, 1991 and Joswig et al., Reproductive Biology and Endocrinology 1:44, 2003.
  • The expression “pregnancy disorders” used herein refers to disorders of the pregnancy in female mammalian, in particular those associated with impaired uterine apoptosis, such as early pregnancy as as disclosed by Schlafke et al., Anat. Rec. 212:47-56, 1985; Parr et al., Biol. Reprod. 36:211-225, 1987; Welsh et al., Am. J. Anat. 192:215-231, 1991 and Joswig et al., Reproductive Biology and Endocrinology 1:44, 2003.
  • For the uses of the invention, an effective amount of a compound of formula (I), or of a pharmaceutically acceptable salt thereof, is administered to the mammals which require such a treatment.
  • The compounds of formula (I) above, and the pharmaceutically acceptable salts thereof, may be used at daily doses of 0.01 to 20 mg per kilo of body weight of the mammal to be treated, preferably at daily doses of from 0.1 to 10 mg/kg. In humans, the dose may vary preferably from 0.5 mg to 1500 mg per day, in particular from 2.5 to 500 mg, depending on the age of the individual to be treated, the type of treatment, prophylactic or curative, and the seriousness of the disorder. The compounds of formula (I) are generally administered as a dosage unit of 0.1 to 500 mg, preferably of 0.5 to 100 mg, of active principle, one to five times a day.
  • Said dosage units are preferably formulated in pharmaceutical compositions in which the active principle is mixed with a pharmaceutical excipient.
  • Thus, according to another of its aspects, the present invention relates to pharmaceutical compositions containing, as an active principle, a compound of formula (I) above or a pharmaceutically acceptable salt thereof.
  • In the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, topical, transdermal or rectal administration, the active ingredients of formula (I) above, and the pharmaceutically acceptable salts thereof, may be administered in unit administration forms, mixed with conventional pharmaceutical supports, to animals and humans for treating the abovementioned disorders. The unit administration forms which are suitable comprise oral forms such as tablets, gel capsules, powders, granules and oral solutions or suspensions, sublingual and buccal administration forms, subcutaneous, intramuscular or intravenous administration forms, local administration forms and rectal administration forms.
  • When a solid composition in the form of tablets is prepared, the main active ingredient is mixed with a pharmaceutical vehicle such as gelatin, starch, lactose, magnesium stearate, talc, gum arabic or the like. The tablets may be coated with sucrose or other suitable materials, or they may be treated such that they have sustained or delayed activity and that they release, in a continuous manner, a predetermined amount of active principle.
  • A preparation of gel capsules is obtained by mixing the active ingredient with a diluent and pouring the mixture obtained into soft or hard gel capsules.
  • A preparation in the form of a syrup or elixir may contain the active ingredient together with a sweetener, preferably a calorie-free sweetener, methylparaben and propylparaben as antiseptics, and also a flavour enhancer and a suitable colorant.
  • The water-dispersible powders or granules may contain the active ingredient mixed with dispersing agents or wetting agents, or suspending agents, such as polyvinylpyrrolidone, and also with sweeteners or flavour correctors.
  • For local administration, the active principle is mixed into an excipient for preparing creams or ointments, or it is dissolved in a vehicle for intraocular administration, for example in the form of an eyewash.
  • For rectal administration, use is made of suppositories prepared with binders which melt at rectal temperature, for example cocoa butter or polyethylene glycols.
  • For parenteral administration, aqueous suspensions, saline solutions or injectable sterile solutions which contain pharmacologically compatible dispersion agents and/or wetting agents, for example propylene glycol or butylene glycol, are used.
  • The active principle may also be formulated in the form of microcapsules, optionally with one or more supports or additives.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 represents Western Blot Analysis with semi-quantification showing the expression of cleaved caspase-3 protein. CTRL=control; LPS=10 μg/ml LPS, 48 h; CA=Chorioamnionitis. Experiments were performed in myometrial samples obtained from 3 different women with Chorioamnionitis (CA group), and with 5 myometrium, obtained at cesarean delivery from 5 different women with uncomplicated pregnancies and stimulated, or not, with LPS (LPS and CTRL groups respectively). *P<0.05 vs. controls.
  • FIG. 2 illustrates Western Blot Analysis with semi-quantification showing the expression of cleaved caspase-3 protein. Controls=time-matched experiments; LPS=10 μg/ml LPS 48 h, TNFα-ab=10 μg/ml LPS+0.6 μg/ml TNFα-ab, 48 h. Experiments were performed on three myometrium obtained at elective cesarean delivery from three different women. *P<0.05 vs. LPS.
  • FIG. 3 illustrates Western Blot Analysis with semi-quantification showing the expression of cleaved caspase-3 protein. Controls=time-matched experiments; LPS=10 μg/ml LPS 48 h. Compound A (10−7M, 10−6M, 10−5M) was added 20 min. prior to LPS stimulation. Experiments were performed on five myometrium, obtained at elective cesarean delivery from five different women. #P<0.05 vs controls, *P<0.05 vs. LPS.
  • FIG. 4 illustrates Western Blot Analysis with semi-quantification showing the expression of BAX and Bcl-2 protein. Controls=time-matched experiments; LPS=10 μg/ml LPS 48 h. Compound A (10−7M, 10−6M, 10−5M) was added 20 min. prior to LPS stimulation. Experiments were performed on seven myometrium, obtained at elective cesarean delivery, from seven different women. #P<0.05 vs controls, *P<0.05 vs. LPS, **P<0.01 vs LPS.
  • FIG. 5 show the effect of Compound A on the concentrations of IL-6 and IL-8. Results (mean±s.e.m, in pg/ml) are from five experiments performed in duplicate in myometrium, obtained at elective cesarean delivery, from five different women. #P<0.05 vs controls, *P<0.05 vs LPS, **P<0.01 vs LPS.
  • The following examples further illustrate the present invention:
  • EXAMPLES Drugs and Solutions
  • Lipopolysaccharide (LPS) (Escherichia coli 055:B5, ref: L2880) was purchased from Sigma-Aldrich and was dissolved in distilled water. Compound A was a gift from Sanofi-Midy Research Center, Exploratory Research Department, Sanofi-Aventis S.p.A. It was dissolved in a mixture of absolute ethanol 30%, dimethylsulfoxide 2%, and distilled water for the 1 mM solution and thereafter diluted in distilled water. The final maximal bath concentration was 0.3% for ethanol and 0.02% for DMSO.
  • Biological Samples
  • Myometrial biopsies were obtained from women during pregnancy in four different clinical situations as outlined: i) a woman with established chorioamnionitis undergoing postpartum hysterectomy; ii) a woman undergoing postpartum hysterectomy for postpartum hemorrhage in the absence of chorioamnionitis; iii) 3 women undergoing elective caesarean section with established chorioamnionitis; iv) 10 women undergoing elective caesarean section for other reasons (cephalo-pelvic disproportion) in the absence of chorioamnionitis. For women undergoing caesarean section, the procedures were all performed prior to the onset of labor at a gestation period between 38 and 40 weeks of pregnancy. Clinical chorioamnionitis was defined classically (Redline R W et al. Placenta 2005; 26 Suppl A: S114-117) as the presence of uterine tenderness and/or purulent or foul-smelling amniotic fluid with any 2 of the following: antepartum temperature of 37.8° C. or more, maternal tachycardia (more than 120 beats/min), maternal leukocytosis more than 18,000 cells/mm3, or fetal tachycardia (more than 160 beats/min) and was confirmed, in all cases included in the present study, by either a positive culture of the placenta or an histological assessment of the placenta by a single pathologist using validated criteria (Redline R W et al. Pediatr Dev Pathol 2003; 6: 435-448). Therefore in the rest of this study chorioamnionitis refers to confirmed chorioamnionitis.
  • Myometrial strips were excised from an immediately subserosal site where the majority of the fibers are in a longitudinal orientation, at an antiplacental site, as previously described (Rouget C, et al. J Clin Endocrinol Metab 2005; 90: 1644-1650, Leroy M J, et al. Biochem Pharmacol 1989; 38: 9-15). This study was approved by the “Comité Consultatif de Protection des Personnes pour la Recherche Biomédicale” (CCPPRB, Dijon, France) and informed consent was obtained from all donors.
    Tissues obtained from women with confirmed chorioamnionitis or postpartum hemorrhage, were either used fresh to perform western blot experiments, or embedded in paraffin for histological assessment and Hoechst staining, as described below.
    Myometrial biopsies obtained from uncomplicated pregnancies were used to develop the LPS experimental model mimicking the effects of chorioamnionitis.
  • Stimulation of Myometrial Biopsies by E. Coli LPS
  • Myometrial biopsies obtained from women with uncomplicated pregnancies were immediately transferred in sterile Dubelcco's Modified Eagle Medium (DMEM) and washed twice with sterile Phosphate Buffer Saline (PBS). Biopsies were cut into small strips, each being placed in a 24-well plate containing 2 ml of culture medium DMEM, without the use of antibiotics. Strips were then incubated at 37° C. with 5% CO2 for 48 h in order to allow cytokine levels to return to basal values (Fortunato S J, et al. Am J Reprod Immunol 1994; 32: 184-187).
  • In order to reproduce with LPS (Escherichia coli 055:B5) the findings on myometrial samples obtained from women with chorioamnionitis, myometrial strips were incubated with three different concentrations of LPS (50 ng/ml, 1 μg/ml and 10 μg/ml) at three different times (8 h, 24 h, 48 h).
    In a second set of experiments, in order to assess the role of TNF in LPS-induced apoptosis in myometrial tissue, the strips were incubated with LPS 10 μg/ml for 48 h with or without anti TNF antibodies (0.6 μg/ml) (Human TNF alpha/TNFSF1A antibody, R&D systems Europe, Lille, France, the concentration of the antibody used was chosen based on the manufacturer's recommendations).
    In a third set of experiments aimed to assess the ability of ADRB3 stimulation to oppose LPS-induced apoptosis, myometrial strips were incubated with LPS 10 μg/ml for 48 h in the presence or absence of Compound A, a selective ADRB3 agonist (Croci T et Al. J Pharmacol Exp Ther 2007; 321: 1118-1126) (0.1 μM, 1 μM, and 10 μM) added immediately prior to begin LPS stimulation. Time-matched control experiments were performed with the solvent of Compound A, i.e. distilled water containing ethanol 0.3% and DMSO 0.02% as final bath concentration, both non-stimulated and LPS-stimulated myometrial samples.
  • At the end of the stimulation period, the supernatants samples and tissues were quickly frozen in liquid nitrogen and stored at −80° C.
  • Western Blotting Analysis
  • Snap-frozen myometrial tissues were homogenized with Ultra-Turrax in homogenization buffer [10 mM Tris-HCl (pH 7.4), 1 mM EDTA, 40 mg/ml−1 leupeptine, 2 mM Pefabloc]. After an initial centrifugation at 500×g for 15 min at 4° C., total protein supernatant content was determined by the Bradford method with BSA as standard. Samples (40 μg of protein by lane) were dissolved (vol/vol) in 2× Laemmli buffer (Laemmli U K et al. J Mol Biol 1970; 47: 69-85) and boiled for 5 min before electrophoresis on a 10% SDS-PAGE. Proteins were transferred to a nitrocellulose membrane (Hybond-P, Amersham Biosciences). In order to block non specific antibody binding, membranes were incubated for 1 h in 10% nonfat dried milk powder in Tris-buffered saline/Tween 20 (TBST) (10 mM Tris, 150 mM NaCl, and 0.1% Tween 20, pH 7.8) at room temperature. Blocked membranes were washed 3 times with TBST. The blots were then incubated overnight at 4° C. with a 1:200 dilution of primary cleaved caspase-3 antibody (ASP175, Cell Signaling Technologies, Beverly, Mass., USA) or a 1:500 dilution of primary BAX polyclonal antibody (sc-493, Santa Cruz, USA) or a 1:500 dilution of primary BCL2 polyclonal antibody (sc-7382, Santa Cruz, USA) in 1% non fat dried milk powder in PBST. After three washes with PBST, the blots were incubated for 45 min with horseradish peroxidase-conjugated antirabbit IgG (NA 934, Amersham, USA) or antimouse IgG (NA 931, Amersham, USA) whole antibody at a dilution of 1:10000 at room temperature and thereafter washed five times with PBST. Immunoreactive proteins were detected by chemiluminescence (ECL™ detection reagents, RPN2105, Amersham, USA) and exposure to a X-ray film (Hyperfilm™, Amersham Bioscience, USA). The intensities of the bands were analyzed densitometrically using the NIH Image 1.62 program and normalized with intensity of bands obtained with monoclonal antibody to Glyceraldehyde 3-Phosphate Dehydrogenase (GAPDH) used as protein loading control. Results are expressed as the mean±s.e.m. in Arbitrary Density Unit (A.D.U).
  • Immunohistochemical Analysis
  • The myometrial strips were fixed for 1 h with paraformaldehyde 4%, then embedded in paraffin and cut into 5-micrometer-thick sections. After deparaffinization of myometrial sections, and rehydratation, antigen retrieval was performed by incubating slides 10 min in warm citric acid buffer pH 6 with a pressure cooker. After endogenous peroxidase activity was blocked with hydrogen peroxide (H2O2) 3%, slides were incubated either with primary anti-cleaved caspase-3 IgG (1:100), washed three times with phosphate buffered saline (PBS) and then incubated with biotinylated anti-rabbit (1:600) immunoglobulin respectively for 1 h. After a new washing, slides were incubated in peroxidase labelled streptavidin (1:800) for 30 min and then with 3-amino-9-ethyl carbazole (AEC) solution until a clearly visible color was developed. The reaction was stopped by extensive washing in double distilled water. Subsequently, the preparations were counterstained with hematoxilin. The histological changes were evaluated in term of variation of global intensity of staining. Negative controls were carried out by omitting the primary antibody.
  • Hoechst Staining
  • To distinguish between apoptotic and necrotic cells, chromatin condensation state was assessed by staining of nuclei with Hoechst-33342. Paraffin embedded sections of myometrium samples were re-hydrated as described above. Slides were then incubated for 2 min with Hoechst-33342 (2 μg/ml). The slides were rinsed with distilled water and mounted with Aquatex©. The stained nuclei were visualized with convert fluorescent microscope at a magnification of ×100, using excitation light at 350 nm.
  • Real Time Quantitative RT-PCR
  • Total RNA was prepared from 5 myometrial tissues obtained from 5 different women, using Trizol solution (Life Technologies, Groningen, The Netherlands) according to the manufacturer's instructions. The integrity of RNA was verified by edithidium bromide staining of agarose gel analysis and by an optical density (OD) absorption ratio OD260 nm/OD280 nm≧1.8. One microgram of total RNA was reverse transcribed with Super script II RNAase H-reverse transcriptase (Invitrogen Life Technologies, Groningen, The Netherlands) using oligo (dT) according to the manufacturer's instruction. Real time quantitative PCR analyses were performed using 25 ng of reverse transcribed total RNA with 200 nM of both sense and anti sense primers in a final volume of 25 μl using the SYBR Green PCR jumpstart reagent (Sigma, Saint Louis, Mo., USA) in an iCycler iQ real time detection system instrument (Bio-Rad, Marnes-1a-Coquette, France). PCR products were also analyzed on edithium bromide stained agarose gel to ensure that a single amplicon of the expected size was indeed obtained.
  • Each reaction was performed in duplicate and Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used in each experiment to control for variability in the quantities of cDNA. Relative quantification for any given gene, expressed as fold variation over control, was calculated after determination of the difference between cycle threshold (Ct) value of the given gene according to manufacturers protocol using the formula 2−Δ(ΔCtA-ΔCtB), in which ΔCtA is ΔCt of the gene of interest; ΔCtB is ΔCt of GAPDH. ΔCt=Ct of the experimental group—Ct of the control group. PCR was performed using specific primers: GAPDH (Forward, 5′TGCACCACCAACTGCTTAGC3′ (SEQ ID NO:1), and Reverse, 5′GGCATGGACTGTGGTC-ATGAG3′ (SEQ ID NO:2)) and CASPASE-3 (Forward, 5′ AGAACTGGACTGTGGCATTGAG 3′ (SEQ ID NO:3), and Reverse, 5′ GCTTGTCGGCATACTGTTTCAG 3′ (SEQ ID NO:4)).
  • Cytokines Level Measurement
  • Cytokines measurements were determined by Cytometric Bead Array (CBA; Bender MedSystems). Supernatants of 5 myometrial strips were incubated with labeled capture beads and detection reagent for 3 h in the dark at room temperature, and analyzed with a flow cytometer (FACSCalibur; BD Biosciences) by using the respective CBA Analysis software (BD Biosciences) and Bender MedSystems software. Cytokine standards for quantification (pg/mL) as well as the blanks were handled in the same manner as the samples. Each experiment was performed in duplicate to ensure for reproducibility of results.
  • Statistical Analysis of Results
  • Differences among groups were determined by analysis of variance (ANOVA) followed by the Dunn's multiple comparison test. Statistical analysis was carried out using GraphPad Instat version 3 (GraphPad Software, San Diego, Calif.). All differences were considered significant when P<0.05.
  • Effect of the Selective ADRB3 Agonist Compound A on Apoptosis and Inflammation
  • Immunostaining of 4 myometrial tissues samples obtained from 4 different women with confirmed chorioamnionitis, using cleaved caspase-3 antibody, showed an intense staining (data not shown) that was observed neither in one control tissue obtained from a woman with postpartum hemorrhage (Sheiner E et al. J Matern Fetal Neonatal Med 2005; 18: 149-154) (data not shown) nor in 3 tissues obtained at elective cesarean delivery (data not shown). This staining was specifically located in myometrial cells. LPS stimulation in optimal conditions (i.e 10 μg/ml for 48 h, determined after time course—8 h, 24 h, 48 h—and dose response—50 ng/ml, 1 μg/ml, 10 μg/ml—experiments of LPS stimulation), was also associated with an intense cleaved caspase-3 staining specifically located in myometrial cells (data not shown).
  • Hoechst-33342 staining of myometrial section obtained from women with chorioamnionitis (n=4), postpartum hemorrhage (n=1) or elective cesarean delivery stimulated (n=4) or not (n=4) with LPS, showed that chorioamnionitis and LPS stimulation (data not shown), were both associated with an increased proportion of cells with condensed chromatin, indicating apoptosis initiation, compared with postpartum hemorrhage or cesarean delivery (data not shown).
  • Western blotting of membranes prepared from pregnant myometrium obtained from the same 3 women with chorioamnionitis or from 5 cesarean section and stimulated, or not (controls), with LPS, revealed a 17-kDa and a 19-kDa bands corresponding to cleaved caspase-3 (data not shown). Densitometric immunoblot analysis indicated that cleaved caspase-3 protein was significantly overexpressed both in case of chorioamnionitis and after LPS stimulation compared with elective cesarean delivery without LPS stimulation used as control (expressed in arbitrary density unit, ADU, 921±39, 941.6±134, vs 452±50.6, for chorioamnionitis, LPS 10 μg/ml and control group respectively, P<0.05) (FIG. 1). The overexpression of cleaved caspase-3 induced by LPS stimulation was in the same extent as that observed in western blot experiments performed in membranes prepared from pregnant myometrium women with chorioamnionitis (FIG. 1) providing external validation of in vitro LPS induced model of chorioamnionitis.
  • This over-expression of cleaved caspase-3 was strongly antagonized by the blockade of TNF-alpha receptor with a selective TNF-alpha antibody suggesting that LPS-induced apoptosis in human near-term myometrium involves TNF-alpha signaling pathway (FIG. 2, ANOVA P<0.05, n=3)
  • Our model of inflammation was validated by the measurement of supernatant cytokines production. In our experimental conditions, LPS stimulation was associated, at 48 h, with a significant increase of IL6 (mean±sem in pg/ml 28860±5257 vs 61860±12190, for control and LPS groups respectively; n=10 experiments from 5 different women, P<0.05) and IL8 (in pg/ml 8855±1486 vs. 16080±2834, for control and LPS groups respectively; n=10 experiments from 5 different women, P<0.05) but not IL1B, IL10, IL12B or TNF levels.
    This over expression of cleaved caspase-3 protein was also observed at a transcriptional level since quantitative real-time RT-PCR showed that, compared with time matched controls, incubation with 10 μg/ml LPS for 48 hours was associated with a 2.36±0.22 fold increased in transcript levels (n=3 experiments performed with myometrial tissues obtained from 3 different women).
  • This weak effect on caspase-3 mRNA expression might be explained by our experimental conditions, i.e. 48 hours of incubation with LPS. Indeed, in an exploratory analysis we assessed time-trend for caspase-3 mRNA expression (3, 6, 12, 48 and 72 hours) and found that the peak for mRNA expression occurred around 3 hours of stimulation (in fold increase compared with controls: 8.6, 6.1, 2.3, 2.1, 1.2 at 3 h, 6 h, 16 h, 48 h and 72 h respectively).
  • In a further set of experiments, western blot experiments revealed that the selective ADRB3 agonist, Compound A, was able to significantly antagonize LPS-induced changes in cleaved caspase-3 expression in a concentration dependent manner (FIG. 3, ANOVA P<0.001,). Furthermore, we observed that LPS-induced activation of the mitochondrial pathway of apoptosis, as expressed by BAX and BCL2 protein up- and down-regulation, respectively, was antagonized in a concentration dependant manner (FIG. 4, ANOVA, P<0.01). Compound A had no effect by itself on cleaved caspase-3, BAX and BCL2 expression in tissues not stimulated with LPS (data not shown). The solvent for the highest concentration of Compound A, i.e. distilled water containing ethanol 0.3% and DMSO 0.02%, had no effect on cleaved caspase-3, BAX and BCL2 expression in tissues either not stimulated or stimulated with LPS (data not shown).
  • The effect of Compound A on caspase-3 over-expression was, at least partially, explained at a transcriptional level since quantitative real-time RT-PCR, showed that ADRB3 treatment was associated with a decreased level of caspase-3 transcripts (in fold increase, normalized to GAPDH, compared with controls 2.08±0.63, 1.17±0.81, 0.84±0.53, 0.93±0.41, respectively for LPS alone and with Compound A 10−7, 10−6 and 10−5 M respectively, ANOVA, P<0.05).
  • Finally, Compound A decreased IL6 and IL8 in a concentration dependent manner, even though the effect was statically significant only for IL8 (ANOVA P=0.01; FIG. 5). Compound A had not effect by itself on IL6 and IL8 release in tissues not stimulated with LPS (data not shown). TNF was under the lower level of detection for the flow-cytometry technique at 48 hours.
  • Altogether, these results demonstrate that Compound A reverses an LPS-induced apoptosis and cytokines production in human near term myometrium.
  • Pharmaceutical Composition According to the Invention
  • As a representative example, a unitary dosage form of a compound of the invention in the form of a tablet may comprise the following constituents:
  • Compound A 50.0 mg
    Mannitol 223.75 mg 
    Sodium Croscaramellose  6.0 mg
    Corn starch 15.0 mg
    Hydroxypropyl-methylcellulose 2.25 mg
    Magnesium stearate  3.0 mg

Claims (18)

1. A method of preventing apoptosis in pregnancy related tissues in a mammalian patient, said method comprising administering to said patient a therapeutically effective amount of a beta-3 adrenergic receptor agonist compound of formula (I):
Figure US20100311829A1-20101209-C00003
wherein:
A is a group of formula (a) or (b):
Figure US20100311829A1-20101209-C00004
where:
R represents a hydrogen or halogen atom, an —S(O)z(C1-C4)alkyl group, an —NHSO2(C1-C4)alkyl group, an —SO2NH(C1-C4)alkyl group, an —NHSO2-phenyl-(C1-C4)alkyl group or an —NHSO2-phenyl group, said phenyl possibly being substituted with a halogen atom, with a (C1-C4)alkyl group or with a (C1-C4)alkoxy group;
R1 represents a hydrogen atom or a (C1-C4)alkyl group, a —CO(C1-C4)alkyl group, a phenyl-(C1-C4)alkyl group or a —CO-phenyl group, said phenyl possibly being substituted with a halogen atom or with a (C1-C4)alkoxy group;
R2 is a hydrogen atom, an —SO2(C1-C4)alkyl group, an —SO2-phenyl-(C1-C4)alkyl group or an —SO2-phenyl group;
X completes a ring of 5 to 8 atoms, said ring being saturated or unsaturated, possibly being substituted with one or two (C1-C4)alkyl groups and bearing one or two carbonyl groups;
n, m and z are, independently, 0, 1 or 2;
R3 represents a hydrogen or halogen atom, a (C1-C6)alkyl group, a (C1-C4)alkoxy group, a —COO(C1-C4)alkyl group, a —CO(C1-C4)alkyl group, an —NHSO2(C1-C4)alkyl group, an —NHSO2-phenyl-(C1-C4)alkyl group, —NO2, —CN, —CONR4R5, —COOH, or a 4,5-dihydro-1,3-oxazol-2-yl or 4,4-dimethyl-4,5-dihydro-1,3-oxazol-2-yl group; and
R4 and R5 represent, independently, a hydrogen atom, a phenyl, a (C1-C4)alkyl group or a phenyl-(C1-C4)alkyl group; or R4 and R5, together with the nitrogen atom to which they are attached, form a ring of 5 to 7 atoms;
or a stereoisomer or mixture of stereoisomers of said beta-3 adrenergic receptor agonist compound;
or a pharmaceutically acceptable salt of said beta-3 adrenergic receptor agonist compound or of said stereoisomer or mixture of stereoisomers thereof.
2. The method according to claim 1, wherein said method prevents or treats a disease or disorder related to apoptosis in a pregnancy related tissue.
3. The method according to claim 2, wherein said pregnancy related tissue is uterine tissue.
4. The method according to claim 2, wherein said disease or disorder is selected from a sexual cycle disorder, a fertility disorder, and a pregnancy disorder.
5. The method according to claim 2, wherein said disease or disorder arises in a patient suffering from genital tract infection.
6. The method according to claim 5, wherein said disease or disorder is preterm labor.
7. The method according to claim 5 or 6 wherein said genital tract infection is chorioamnionitis.
8. The method according to claim 1, wherein said patient is a pregnant woman.
9. The method according to claim 2, wherein said disease or disorder comprises intrauterine growth restriction or preeclampsia.
10. The method according to claim 2, wherein said disease or disorder comprises preterm premature rupture of fetal membranes.
11. The method according to claim 1, wherein n and m are each zero.
12. The method according to claim 1, wherein R1 is a hydrogen atom.
13. The method according to claim 1, wherein R is an —NHSO2(C1-C4)alkyl group, an —NHSO2-phenyl-(C1-C4)alkyl group or an —NHSO2-phenyl group.
14. The method according to claim 1, wherein R3 is —COO(C1-C4)alkyl, —CO(C1-C4)alkyl, or CONR4R5.
15. The method according to claim 1, wherein R3 is in position 4 of the benzene.
16. The method according to claim 1, wherein z is 2.
17. The method according to claim 1, wherein X is methylene, ethylene or propylene.
18. The method according to claim 1, wherein X is a carbonyl, a —CO—CO-group, a —CO—C((C1-C4)alkyl)2-CO— group, a methylene mono-substituted or disubstituted with (C1-C4)alkyl or a —COCH2— group.
US12/652,317 2007-07-05 2010-01-05 Therapeutic uses of beta-3 adrenergic receptor agonist derivatives in particular to modulate apoptosis Expired - Fee Related US9161925B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP07290845A EP2011490A1 (en) 2007-07-05 2007-07-05 Beta adrenergic receptor ligand derivatives for modulating apoptosis
EP07290845.2 2007-07-05
EP07290845 2007-07-05
PCT/IB2008/002752 WO2009019607A2 (en) 2007-07-05 2008-07-04 Beta adrenergic receptor ligand derivatives for modulating apoptosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2008/002752 Continuation WO2009019607A2 (en) 2007-07-05 2008-07-04 Beta adrenergic receptor ligand derivatives for modulating apoptosis

Publications (2)

Publication Number Publication Date
US20100311829A1 true US20100311829A1 (en) 2010-12-09
US9161925B2 US9161925B2 (en) 2015-10-20

Family

ID=38519604

Family Applications (1)

Application Number Title Priority Date Filing Date
US12/652,317 Expired - Fee Related US9161925B2 (en) 2007-07-05 2010-01-05 Therapeutic uses of beta-3 adrenergic receptor agonist derivatives in particular to modulate apoptosis

Country Status (4)

Country Link
US (1) US9161925B2 (en)
EP (2) EP2011490A1 (en)
JP (1) JP5539195B2 (en)
WO (1) WO2009019607A2 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009061918A1 (en) 2007-11-06 2009-05-14 Oklahoma Medical Research Foundation Extracellular histones as biomarkers for prognosis and molecular targets for therapy
CN102764438A (en) * 2012-01-30 2012-11-07 林曙光 Novel application of beta3 adrenoceptor agonist

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2817257B1 (en) * 2000-11-30 2009-03-20 Sanofi Synthelabo CYCLOHEXYL (ALKYL) -PROPANOLAMINES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2840301B1 (en) * 2002-05-29 2007-03-23 Sanofi Synthelabo PHENYL-CYCLOHEXYL-PROPANOLAMINE DERIVATIVES, THEIR PREPARATION AND THEIR USE IN THERAPEUTICS
FR2840304B1 (en) * 2002-05-29 2007-05-18 Sanofi Synthelabo OXOPHENYL-CYCLOHEXYL-PROPANOLAMINE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Ngassa et al (Int J Gynaecol Obstet 47:241-246, 1994 (Abstract only)) *
Perfettini et al (Infection and Immunity 68:2237-2244, 2000) *

Also Published As

Publication number Publication date
EP2011490A1 (en) 2009-01-07
JP2010532342A (en) 2010-10-07
WO2009019607A2 (en) 2009-02-12
EP2173335A2 (en) 2010-04-14
US9161925B2 (en) 2015-10-20
WO2009019607A4 (en) 2009-05-28
WO2009019607A3 (en) 2009-04-09
EP2173335B1 (en) 2012-09-19
JP5539195B2 (en) 2014-07-02

Similar Documents

Publication Publication Date Title
Oh et al. Autophagy-related proteins, LC3 and Beclin-1, in placentas from pregnancies complicated by preeclampsia
US20090281110A1 (en) Method of Treatment
WO2013052995A2 (en) Improved developmental competence of oocytes
CA2837359C (en) Methods and compositions for enhancing fertility and/or inhibiting pregnancy failure and restoring glucose tolerance
CN106796243B (en) Judge the method for the risk of premature labor and/or infant of low-birth weight&#39;s fertility and the kit for this method
US9161925B2 (en) Therapeutic uses of beta-3 adrenergic receptor agonist derivatives in particular to modulate apoptosis
JP2022504990A (en) Compositions and Methods for Treating Vascular Ehlers-Danlos Syndrome and Related Disorders
US20220390474A1 (en) Detection and Treatment of Conditions Related to Luteinizing Hormone/Follicle-Stimulating Hormone (LH/FSH) Levels
KR20140138824A (en) Method for treating gynecological diseases
KR102675587B1 (en) Pharmaceutical composition for preventing or treating diseases related to abnormal proliferation of endometrial cells or trophoblast cells comprising fenbendazole, oxibendazole or mixture thereof
US20160279202A1 (en) Interleukin-1 (il-1) inhibitors for treating fertility disorders
JP6840775B2 (en) Methods to reduce perinatal illness and / or mortality
CZ20011524A3 (en) Use of thiazolidinediones derivatives for preventing uterine contractions in premature labor or lactation
KR102434351B1 (en) Pharmaceutical composition for preventing or treating male reproductive disease comprising butylated hydroxyanisole, butylated hydroxytoluene or mixture thereof
US20070270392A1 (en) Use of a Cyclopentenone Prostaglandin for Delaying the Onset and/or Preventing the Continuation of Labour
KR102066149B1 (en) Composition comprising C-C motif chemokine ligand 2(CCL2) for improving pregnancy
US20230285347A1 (en) Preterm Labour with Prostaglandin E2 Receptor Agonists
CN115590849A (en) Oxytocin antagonist dosing regimen to promote embryo implantation and prevent miscarriage
US20190255025A1 (en) Methods and compositions for enhancing fertility and/or inhibiting pregnancy failure and restoring glucose tolerance
Nakashima et al. The Role of Autophagy in Maintaining Pregnancy
KR20220075340A (en) Oxytocin antagonist administration regimen to promote embryo implantation and prevent miscarriage
WO2022200848A1 (en) Pterostilbene for use in the treatment or prevention of infertility
Coughlan et al. Polycystic ovarian syndrome
EP1827451A1 (en) Use of a cyclopentenone prostaglandin for delaying for the onset and/or preventing the continuation of labour
Konje Ectopic Pregnancy Is Associated with High Anandamide Levels and Aberrant Expression of FAAH and CB1 in Fallopian Tubes

Legal Events

Date Code Title Description
AS Assignment

Owner name: SANOFI-AVENTIS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BARDOU, MARC;REEL/FRAME:024846/0169

Effective date: 20100615

AS Assignment

Owner name: SANOFI, FRANCE

Free format text: CHANGE OF NAME;ASSIGNOR:SANOFI-AVENTIS;REEL/FRAME:028413/0927

Effective date: 20110511

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20231020